#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Male-Biased Aganglionic Megacolon in the TashT Mouse Line Due to Perturbation of Silencer Elements in a Large Gene Desert of Chromosome 10


Hirschsprung’s disease (also known as aganglionic megacolon) is a severe congenital defect of the enteric nervous system (ENS) resulting in complete failure to pass stools. It is characterized by the absence of neural ganglia (aganglionosis) in the distal gut due to incomplete colonization of the embryonic intestines by neural crest cells (NCC), the ENS precursors. Hirschsprung’s disease has an incidence of 1 in 5000 newborns and a 4:1 male sex bias. Although many genes have been associated with this complex genetic disease, most of its heritability as well as its male sex bias remain unexplained. Here, we describe an insertional mutant mouse line (“TashT”) in which virtually all homozygotes display colonic aganglionosis due to defective migration of enteric NCC, but in which only a subset of homozygotes develops megacolon. Surprisingly, this group is almost exclusively male. The TashT ENS defect stems, at least in part, from the disruption of long-range interactions between evolutionarily conserved elements with silencer activity and Fam162b, resulting in NCC-specific upregulation of this uncharacterized protein coding gene. Global analysis of gene expression further revealed that several hundreds of genes are significantly deregulated in TashT enteric NCC. Interestingly, this dataset includes multiple X-linked candidate genes potentially underlying the male sex bias. Taken together, our data pave the way for a clearer understanding of the intriguing male sex bias of Hirschsprung’s disease.


Vyšlo v časopise: Male-Biased Aganglionic Megacolon in the TashT Mouse Line Due to Perturbation of Silencer Elements in a Large Gene Desert of Chromosome 10. PLoS Genet 11(3): e32767. doi:10.1371/journal.pgen.1005093
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005093

Souhrn

Hirschsprung’s disease (also known as aganglionic megacolon) is a severe congenital defect of the enteric nervous system (ENS) resulting in complete failure to pass stools. It is characterized by the absence of neural ganglia (aganglionosis) in the distal gut due to incomplete colonization of the embryonic intestines by neural crest cells (NCC), the ENS precursors. Hirschsprung’s disease has an incidence of 1 in 5000 newborns and a 4:1 male sex bias. Although many genes have been associated with this complex genetic disease, most of its heritability as well as its male sex bias remain unexplained. Here, we describe an insertional mutant mouse line (“TashT”) in which virtually all homozygotes display colonic aganglionosis due to defective migration of enteric NCC, but in which only a subset of homozygotes develops megacolon. Surprisingly, this group is almost exclusively male. The TashT ENS defect stems, at least in part, from the disruption of long-range interactions between evolutionarily conserved elements with silencer activity and Fam162b, resulting in NCC-specific upregulation of this uncharacterized protein coding gene. Global analysis of gene expression further revealed that several hundreds of genes are significantly deregulated in TashT enteric NCC. Interestingly, this dataset includes multiple X-linked candidate genes potentially underlying the male sex bias. Taken together, our data pave the way for a clearer understanding of the intriguing male sex bias of Hirschsprung’s disease.


Zdroje

1. Bergeron KF, Silversides DW, Pilon N (2013) The developmental genetics of Hirschsprung's disease. Clin Genet 83: 15–22. doi: 10.1111/cge.12032 23043324

2. Anderson RB, Newgreen DF, Young HM (2006) Neural crest and the development of the enteric nervous system. Adv Exp Med Biol 589: 181–196. 17076282

3. Nishiyama C, Uesaka T, Manabe T, Yonekura Y, Nagasawa T, et al. (2012) Trans-mesenteric neural crest cells are the principal source of the colonic enteric nervous system. Nat Neurosci 15: 1211–1218. doi: 10.1038/nn.3184 22902718

4. Burns AJ, Champeval D, Le Douarin NM (2000) Sacral neural crest cells colonise aganglionic hindgut in vivo but fail to compensate for lack of enteric ganglia. Dev Biol 219: 30–43. 10677253

5. Amiel J, Sproat-Emison E, Garcia-Barcelo M, Lantieri F, Burzynski G, et al. (2008) Hirschsprung disease, associated syndromes and genetics: a review. J Med Genet 45: 1–14. 17965226

6. Alves MM, Sribudiani Y, Brouwer RW, Amiel J, Antinolo G, et al. (2013) Contribution of rare and common variants determine complex diseases-Hirschsprung disease as a model. Dev Biol 382: 320–329. doi: 10.1016/j.ydbio.2013.05.019 23707863

7. Obermayr F, Hotta R, Enomoto H, Young HM (2013) Development and developmental disorders of the enteric nervous system. Nat Rev Gastroenterol Hepatol 10: 43–57. doi: 10.1038/nrgastro.2012.234 23229326

8. Hosoda K, Hammer RE, Richardson JA, Baynash AG, Cheung JC, et al. (1994) Targeted and natural (piebald-lethal) mutations of endothelin-B receptor gene produce megacolon associated with spotted coat color in mice. Cell 79: 1267–1276. 8001159

9. Baynash AG, Hosoda K, Giaid A, Richardson JA, Emoto N, et al. (1994) Interaction of endothelin-3 with endothelin-B receptor is essential for development of epidermal melanocytes and enteric neurons. Cell 79: 1277–1285. 8001160

10. Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira SA, et al. (1996) Renal agenesis and the absence of enteric neurons in mice lacking GDNF. Nature 382: 70–73. 8657306

11. Asai N, Fukuda T, Wu Z, Enomoto A, Pachnis V, et al. (2006) Targeted mutation of serine 697 in the Ret tyrosine kinase causes migration defect of enteric neural crest cells. Development 133: 4507–4516. 17050626

12. Schuchardt A, D'Agati V, Larsson-Blomberg L, Costantini F, Pachnis V (1994) Defects in the kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret. Nature 367: 380–383. 8114940

13. Uesaka T, Nagashimada M, Yonemura S, Enomoto H (2008) Diminished Ret expression compromises neuronal survival in the colon and causes intestinal aganglionosis in mice. J Clin Invest 118: 1890–1898. doi: 10.1172/JCI34425 18414682

14. McCallion AS, Stames E, Conlon RA, Chakravarti A (2003) Phenotype variation in two-locus mouse models of Hirschsprung disease: tissue-specific interaction between Ret and Ednrb. Proc Natl Acad Sci U S A 100: 1826–1831. 12574515

15. Methot D, Reudelhuber TL, Silversides DW (1995) Evaluation of tyrosinase minigene co-injection as a marker for genetic manipulations in transgenic mice. Nucleic Acids Res 23: 4551–4556. 8524641

16. Boyer A, Pilon N, Raiwet DL, Lussier JG, Silversides DW (2006) Human and pig SRY 5' flanking sequences can direct reporter transgene expression to the genital ridge and to migrating neural crest cells. Dev Dyn 235: 623–632. 16411204

17. Pilon N, Raiwet D, Viger RS, Silversides DW (2008) Novel pre- and post-gastrulation expression of Gata4 within cells of the inner cell mass and migratory neural crest cells. Dev Dyn 237: 1133–1143. doi: 10.1002/dvdy.21496 18351674

18. Young HM, Bergner AJ, Anderson RB, Enomoto H, Milbrandt J, et al. (2004) Dynamics of neural crest-derived cell migration in the embryonic mouse gut. Dev Biol 270: 455–473. 15183726

19. Barlow A, de Graaff E, Pachnis V (2003) Enteric nervous system progenitors are coordinately controlled by the G protein-coupled receptor EDNRB and the receptor tyrosine kinase RET. Neuron 40: 905–916. 14659090

20. Nagy N, Goldstein AM (2006) Endothelin-3 regulates neural crest cell proliferation and differentiation in the hindgut enteric nervous system. Dev Biol 293: 203–217. 16519884

21. Young HM, Hearn CJ, Farlie PG, Canty AJ, Thomas PQ, et al. (2001) GDNF is a chemoattractant for enteric neural cells. Dev Biol 229: 503–516. 11150245

22. Mwizerwa O, Das P, Nagy N, Akbareian SE, Mably JD, et al. (2011) Gdnf is mitogenic, neurotrophic, and chemoattractive to enteric neural crest cells in the embryonic colon. Dev Dyn 240: 1402–1411. doi: 10.1002/dvdy.22630 21465624

23. Bergeron KF, Cardinal T, Pilon N (2013) A quantitative cell migration assay for murine enteric neural progenitors. J Vis Exp: e50709. doi: 10.3791/50709 24084298

24. Gianino S, Grider JR, Cresswell J, Enomoto H, Heuckeroth RO (2003) GDNF availability determines enteric neuron number by controlling precursor proliferation. Development 130: 2187–2198. 12668632

25. Ngan ES, Garcia-Barcelo MM, Yip BH, Poon HC, Lau ST, et al. (2011) Hedgehog/Notch-induced premature gliogenesis represents a new disease mechanism for Hirschsprung disease in mice and humans. J Clin Invest 121: 3467–3478. doi: 10.1172/JCI43737 21841314

26. Wang Z, Cao R, Taylor K, Briley A, Caldwell C, et al. (2013) The properties of genome conformation and spatial gene interaction and regulation networks of normal and malignant human cell types. PLoS One 8: e58793. doi: 10.1371/journal.pone.0058793 23536826

27. Naumova N, Smith EM, Zhan Y, Dekker J (2012) Analysis of long-range chromatin interactions using Chromosome Conformation Capture. Methods 58: 192–203. doi: 10.1016/j.ymeth.2012.07.022 22903059

28. Antonellis A, Huynh JL, Lee-Lin SQ, Vinton RM, Renaud G, et al. (2008) Identification of neural crest and glial enhancers at the mouse Sox10 locus through transgenesis in zebrafish. PLoS Genet 4: e1000174. doi: 10.1371/journal.pgen.1000174 18773071

29. Werner T, Hammer A, Wahlbuhl M, Bosl MR, Wegner M (2007) Multiple conserved regulatory elements with overlapping functions determine Sox10 expression in mouse embryogenesis. Nucleic Acids Res 35: 6526–6538. 17897962

30. Heanue TA, Pachnis V (2006) Expression profiling the developing mammalian enteric nervous system identifies marker and candidate Hirschsprung disease genes. Proc Natl Acad Sci U S A 103: 6919–6924. 16632597

31. Vohra BP, Tsuji K, Nagashimada M, Uesaka T, Wind D, et al. (2006) Differential gene expression and functional analysis implicate novel mechanisms in enteric nervous system precursor migration and neuritogenesis. Dev Biol 298: 259–271. 16904662

32. Akbareian SE, Nagy N, Steiger CE, Mably JD, Miller SA, et al. (2013) Enteric neural crest-derived cells promote their migration by modifying their microenvironment through tenascin-C production. Dev Biol 382: 446–456. doi: 10.1016/j.ydbio.2013.08.006 23958436

33. Brauer PR, Bolender DL, Markwald RR (1985) The distribution and spatial organization of the extracellular matrix encountered by mesencephalic neural crest cells. Anat Rec 211: 57–68. 3985379

34. Druckenbrod NR, Epstein ML (2009) Age-dependent changes in the gut environment restrict the invasion of the hindgut by enteric neural progenitors. Development 136: 3195–3203. doi: 10.1242/dev.031302 19700623

35. Hotta R, Anderson RB, Kobayashi K, Newgreen DF, Young HM (2010) Effects of tissue age, presence of neurones and endothelin-3 on the ability of enteric neurone precursors to colonize recipient gut: implications for cell-based therapies. Neurogastroenterol Motil 22: 331–e386. doi: 10.1111/j.1365-2982.2009.01411.x 19775251

36. Del Rio T, Nishitani AM, Yu WM, Goodrich LV (2013) In vivo analysis of Lrig genes reveals redundant and independent functions in the inner ear. PLoS Genet 9: e1003824. doi: 10.1371/journal.pgen.1003824 24086156

37. Ledda F, Bieraugel O, Fard SS, Vilar M, Paratcha G (2008) Lrig1 is an endogenous inhibitor of Ret receptor tyrosine kinase activation, downstream signaling, and biological responses to GDNF. J Neurosci 28: 39–49. doi: 10.1523/JNEUROSCI.2196-07.2008 18171921

38. Young HM, Bergner AJ, Simpson MJ, McKeown SJ, Hao MM, et al. (2014) Colonizing while migrating: how do individual enteric neural crest cells behave? BMC Biol 12: 23. doi: 10.1186/1741-7007-12-23 24670214

39. Sanyal A, Lajoie BR, Jain G, Dekker J (2012) The long-range interaction landscape of gene promoters. Nature 489: 109–113. doi: 10.1038/nature11279 22955621

40. Li G, Ruan X, Auerbach RK, Sandhu KS, Zheng M, et al. (2012) Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell 148: 84–98. doi: 10.1016/j.cell.2011.12.014 22265404

41. Fanucchi S, Shibayama Y, Burd S, Weinberg MS, Mhlanga MM (2013) Chromosomal contact permits transcription between coregulated genes. Cell 155: 606–620. doi: 10.1016/j.cell.2013.09.051 24243018

42. Feng W, Leach SM, Tipney H, Phang T, Geraci M, et al. (2009) Spatial and temporal analysis of gene expression during growth and fusion of the mouse facial prominences. PLoS One 4: e8066. doi: 10.1371/journal.pone.0008066 20016822

43. Ro S, Hwang SJ, Muto M, Jewett WK, Spencer NJ (2006) Anatomic modifications in the enteric nervous system of piebald mice and physiological consequences to colonic motor activity. Am J Physiol Gastrointest Liver Physiol 290: G710–718. 16339294

44. Cantrell VA, Owens SE, Chandler RL, Airey DC, Bradley KM, et al. (2004) Interactions between Sox10 and EdnrB modulate penetrance and severity of aganglionosis in the Sox10Dom mouse model of Hirschsprung disease. Hum Mol Genet 13: 2289–2301. 15294878

45. Dang R, Torigoe D, Suzuki S, Kikkawa Y, Moritoh K, et al. (2011) Genetic background strongly modifies the severity of symptoms of Hirschsprung disease, but not hearing loss in rats carrying Ednrb(sl) mutations. PLoS ONE 6: e24086. doi: 10.1371/journal.pone.0024086 21915282

46. Vohra BP, Planer W, Armon J, Fu M, Jain S, et al. (2007) Reduced endothelin converting enzyme-1 and endothelin-3 mRNA in the developing bowel of male mice may increase expressivity and penetrance of Hirschsprung disease-like distal intestinal aganglionosis. Dev Dyn 236: 106–117. 17131407

47. Emison ES, McCallion AS, Kashuk CS, Bush RT, Grice E, et al. (2005) A common sex-dependent mutation in a RET enhancer underlies Hirschsprung disease risk. Nature 434: 857–863. 15829955

48. Berletch JB, Yang F, Xu J, Carrel L, Disteche CM (2011) Genes that escape from X inactivation. Hum Genet 130: 237–245. doi: 10.1007/s00439-011-1011-z 21614513

49. Carrel L, Willard HF (2005) X-inactivation profile reveals extensive variability in X-linked gene expression in females. Nature 434: 400–404. 15772666

50. Deng X, Berletch JB, Nguyen DK, Disteche CM (2014) X chromosome regulation: diverse patterns in development, tissues and disease. Nat Rev Genet 15: 367–378. doi: 10.1038/nrg3687 24733023

51. Huynh KD, Lee JT (2003) Inheritance of a pre-inactivated paternal X chromosome in early mouse embryos. Nature 426: 857–862. 14661031

52. Wolstenholme JT, Rissman EF, Bekiranov S (2013) Sexual differentiation in the developing mouse brain: contributions of sex chromosome genes. Genes Brain Behav 12: 166–180. doi: 10.1111/gbb.12010 23210685

53. Xu J, Burgoyne PS, Arnold AP (2002) Sex differences in sex chromosome gene expression in mouse brain. Hum Mol Genet 11: 1409–1419. 12023983

54. Yamamoto T, Wilsdon A, Joss S, Isidor B, Erlandsson A, et al. (2014) An emerging phenotype of Xq22 microdeletions in females with severe intellectual disability, hypotonia and behavioral abnormalities. J Hum Genet 59: 300–306. doi: 10.1038/jhg.2014.21 24646727

55. Nagy A, Gertsenstein M, Vintersten K, Behringer R (2003) Manipulating the mouse embryo, A laboratory manual, 3rd Edition. Cold spring Harbor, New-York: Cold Spring Harbor Laboratory Press. doi: 10.1007/978-1-60327-019-9_13 19504073

56. Kothary R, Clapoff S, Darling S, Perry MD, Moran LA, et al. (1989) Inducible expression of an hsp68-lacZ hybrid gene in transgenic mice. Development 105: 707–714. 2557196

57. Lu W, Phillips CL, Killen PD, Hlaing T, Harrison WR, et al. (1999) Insertional mutation of the collagen genes Col4a3 and Col4a4 in a mouse model of Alport syndrome. Genomics 61: 113–124. 10534397

58. Matsuda Y, Chapman VM (1995) Application of fluorescence in situ hybridization in genome analysis of the mouse. Electrophoresis 16: 261–272. 7774567

59. Enomoto H, Araki T, Jackman A, Heuckeroth RO, Snider WD, et al. (1998) GFR alpha1-deficient mice have deficits in the enteric nervous system and kidneys. Neuron 21: 317–324. 9728913

60. Abramoff MD, Magalhaes PJ, Ram SJ (2004) Image processing with ImageJ. Biophotonics International 11: 36–42.

61. Nordeen SK (1988) Luciferase reporter gene vectors for analysis of promoters and enhancers. Biotechniques 6: 454–458. 2908509

62. Sanchez-Ferras O, Coutaud B, Djavanbakht Samani T, Tremblay I, Souchkova O, et al. (2012) Caudal-related homeobox (Cdx) protein-dependent integration of canonical Wnt signaling on paired-box 3 (Pax3) neural crest enhancer. J Biol Chem 287: 16623–16635. doi: 10.1074/jbc.M112.356394 22457346

63. Sanchez-Ferras O, Bernas G, Laberge-Perrault E, Pilon N (2014) Induction and dorsal restriction of Paired-box 3 (Pax3) gene expression in the caudal neuroectoderm is mediated by integration of multiple pathways on a short neural crest enhancer. Biochim Biophys Acta 1839: 546–558. doi: 10.1016/j.bbagrm.2014.04.023 24815547

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 3
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#